ArticlePDF Available

Prevention and Control of Chagas Disease – An Overview

Authors:

Abstract and Figures

Chagas disease is the main cause of heart failure and sudden death in the Western Hemisphere. The literature of the last decades reported on the changing epidemiological profiles of Chagas disease, which now threats the human population in the cities. The exodus of the Latin America people to the Northern Hemisphere explains the growing concern in countries where the transmission of Trypanosoma cruzi was accidental or transferred from a mother to her offspring. Herein, we present the evidence of the possible acquisition of the T. cruzi infection by sex. The staggering demonstration of the transmission of the T. cruzi infections from males and females to naïve mates by intercourse introduces substantial changes in the surveillance of the Chagas disease. Notably, the sexual transmission of the T. cruzi introduces changes in the concepts of medical care, prevention and control; specifically, the risk for the vertical transfer of the parasite-induced kDNA mutations, underpinning the genetically driven autoimmunity, inheritance, and pathogenesis associated with multifaceted clinical manifestations of Chagas disease with high ratios of morbidity and mortality. In this regard, the endemics require much paradigm research with new approaches and innovation technologies, aiming at its control. For example, the recent knowledge anticipates useful measures for preventing the potential forthcoming pandemic Chagas parasites. A long-lasting multicenter research program is needed for creative, drug discovery for curtailment of Chagas disease. Meanwhile, the prevention shall rely on the education, information, and communication program for health. Keywords : Trypanosoma cruzi; sexual transmission; pathogenesis; drug development; prevention; education.
Content may be subject to copyright.
_____________________________________________________________________________________________________
*Corresponding author: Email: antonioteixeirarl@gmail.com;
International STD Research
& Reviews
7(2): 1-15, 2018; Article no.ISRR.42594
ISSN: 2347-5196, NLM ID: 101666147
Prevention and Control of Chagas Disease –
An Overview
A. R. L. Teixeira
1*
, C. Gomes
2
, A. C. Rosa
1
, P. F. Araujo
1
, C. E. Anunciação
2
,
E. Silveira-Lacerda
2
, A. B. Almeida
1
and S. Petrofeza
2
1
Chagas Disease Multidisciplinary Research Laboratory, Faculty of Medicine, University of Brasília,
Brazil.
2
Center for Research and Prevention of Neglected Diseases, Institute of Biology, Federal University of
Goiás, Brazil.
Authors’ contributions
All authors had substantial contribution to the literature review, conception of the work, revised the
work critically for important intellectual content, approved the final version and agree to be
accountable for all aspects.
Article Information
DOI: 10.9734/ISRR/2018/42594
Editor(s):
(1) Anjana Verma, Assistant Professor, Department of Community Medicine, Geetanjali Medical College, Geetanjali Medcity,
India.
Reviewers:
(1)
Alejandro Carabarín-Lima, Benemérita Universidad Autónoma de Puebla, México.
(2)
Natalia Indira Vargas Cuentas, Universidad de Ciencias y Humanidades, Peru.
Complete Peer review History:
http://www.sciencedomain.org/review-history/25651
Received 10
th
May 2018
Accepted 19
th
July 2018
Published 24
th
July 2018
ABSTRACT
Chagas disease is the main cause of heart failure and sudden death in the Western Hemisphere.
The literature of the last decades reported on the changing epidemiological profiles of Chagas
disease, which now threats the human population in the cities. The exodus of the Latin America
people to the Northern Hemisphere explains the growing concern in countries where the
transmission of Trypanosoma cruzi was accidental or transferred from a mother to her offspring.
Herein, we present the evidence of the possible acquisition of the T. cruzi infection by sex. The
staggering demonstration of the transmission of the T. cruzi infections from males and females to
naïve mates by intercourse introduces substantial changes in the surveillance of the Chagas
disease. Notably, the sexual transmission of the T. cruzi introduces changes in the concepts of
medical care, prevention and control; specifically, the risk for the vertical transfer of the parasite-
induced kDNA mutations, underpinning the genetically driven autoimmunity, inheritance, and
pathogenesis associated with multifaceted clinical manifestations of Chagas disease with high
Review Article
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
2
ratios of morbidity and mortality. In this regard, the endemics require much paradigm research with
new approaches and innovation technologies, aiming at its control. For example, the recent
knowledge anticipates useful measures for preventing the potential forthcoming pandemic Chagas
parasites. A long-lasting multicenter research program is needed for creative, drug discovery for
curtailment of Chagas disease. Meanwhile, the prevention shall rely on the education, information,
and communication program for health.
Keywords: Trypanosoma cruzi; sexual transmission; pathogenesis; drug development; prevention;
education.
ACHRONYMS
ATPase : adenyltriphosphatase.
Benznidazole and Nifurtimox : nitroderatives used for the treatment of Chagas disease.
CCD : chronic Chagas disease.
CSB : kinetoplast minicircle constant sequence block.
CYP5128–30 : fenarimol herbicide azole inhibitor of T. cruzi.
EICH : education, information, communication for health.
elF2a : elongation factor 2a.
ELISA : enzyme-linked immunosorbent assay.
IFI : immunofluorescence indirect.
ik2 : eukaryote kinase subunit 2.
EMBL : European molecular biology laboratory.
ERV : endogenous retro virus.
LINE-1 : long interspersed nuclear element-1.
MAL-R : long terminal repeats.
NAT : nucleic acid test.
nDNA
: T, cruzi nuclear DNA.
kDNA : T. cruzi mitochondrion kinetoplast DNA;
PI3K : phosphate inositol 3 kinase.
PCR : polymerase chain reaction.
32
P-dATP : P'-(32)P-labeled; 2'-deoxyadenosine triphosphate.
1. INTRODUCTION
The Kinetoplastid flagellates in the Family
Trypanosomatidae (from greek; trypanon, auger;
soma, body) include Trypanosoma cruzi, the
agent of the American trypanosomiasis [1,2]. The
T. cruzi belongs to the Stercorarian group of
flagellates that accomplishes its life cycle in the
gut of the invertebrate host; the insect sucks in
the blood trypomastigote, which transforms into
epimastigote in the foregut and reverts to
infective metacyclic trypomastigote in the hind
gut. The requirement of an invertebrate host for
completion of the protozoa lifecycle ascribes to
the beginning of the T. cruzi enzootic infection of
mammals that dwell in the American Continent
[3-5]. The enzootics include reservoirs of the
orders Marsupialia, Edentata, Chiroptera,
Carnivora, Artiodactyla, Rodentia and Primates,
upon whom triatomine bugs prey and transmit T.
cruzi [5-7]. The spreading of the T. cruzi
infections requires a broad diversity of triatomine
vectors and mammalian hosts [5-10].
In past decades, the epidemiologic profiles of
American trypanosomiasis and Chagas disease
underwent drastic changes due to the rural
exodus and, therefore, now they become an
emerging urban problem [6-18]. The changes of
the profiles follow pattern of the migration of T.
cruzi-infected people from the South America
continent and their settlement in the Northern
Hemisphere [18].
Herein, we review the literature that describes
the recent change of epidemiologic profiles of the
Chagas disease. The review integrates the
available data and information in men and in
laboratory animals with growing concern about
new technologies for preventing Chagas disease
that threatens the people worldwide [7-19]. With
this respect, this review article presents up-to-
date parasitological, clinic, cell biology,
immunologic, genetic, pathology and the scanty
epidemiologic information for the attention of the
scientific community and for the health
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
3
authorities. The aim of this review is to spurring a
long-lasting program, aiming at a new drug to
treat the carrier and to eradicate the endemic T.
cruzi infections and control of the sexual
transmission of the T. cruzi [18-20]. This review
anticipates that the curtailment of Chagas
disease needs international solidarity and
multicenter research groups to undertake
efficacious, creative, prevention paradigm
achievements. In this regard, a mass-media
Education, Information, and Communication for
Health (EICH) program to prevent Chagas
disease in the forthcoming decades can no
longer be neglected.
2. TRANSMISSION ROUTES
The main public health problem initiated,
possibly, with the accidental triatomine bug-
transmission of the T. cruzi infections to human
[21-23]. However, the route for the bug hindgut’s
trypomastigote entry into the host’s body and
parasitic cell growth, is cumbersome: i) in the
absence of systematic information, it is assumed
that most acute T. cruzi infections stem from
triatomine bug’s bite, supposedly, in hinterland
endemic areas; ii) incomplete knowledge about
the bug’s feces T. cruzi contamination ratios in
countless ecotypes of the America continent [24-
30]; iii) triatomine bug’s night habits, frequency of
accomplishing a full meal and ratios of T. cruzi
contamination, is mostly unknown [27-29]; iv)
quick feeding ability to obtaining a full blood
meal, distension of the abdomen and bug
immediate defecation, remains to be determined
[27-29]; v) triatomine bite site scratch-induced
allergic reactions and vase dilation promotes
bug’s proboscis cannulating and suck-in a full
blood meal, defecation and spread of the
excreta, and the T. cruzi contaminates the
human body [27-29]; nonetheless, in the
absence of an allergic reaction and vase dilation
the prey does not aware about the annoyance
that incites the scratching, which renders much
difficult a full completion of the time consuming
blood meal, defecation and possible
contamination of the host’s body [24-31]; vi) the
frequency of the hypersensitivity lesion at the
port of entry of the parasite in the skin
(Chagoma) or in the eye conjunctiva (Romaña’s
sign) is, possibly, less than circa 1 out of 1000
acute infection [5,6].
The spectrum of drawbacks translates an array
of difficulties towards collecting family information
about bug’s transmitted T. cruzi infections to the
human population. However, the early
recognition of the vector-transmitted T. cruzi
infections posited first in the rank among routes
of the T. cruzi contamination of humans [18,23].
In this regard, further investigation is needed to
determine whether the ratios of T. cruzi-positive
triatomine infestations correlate to the prevalence
of Chagas disease in the cities.
The acquisition of the T. cruzi infections per os is
the most ancient route for the T. cruzi
contaminating the insectivorous mammals’
reservoirs in the wilderness [31-33]. A few
epidemiologic studies refer to the outbreaks of
acute Chagas disease in humans, possibly, by
imprudent ingestion of food contaminated with
the triatomine bug’s feces [2,32,33]. Next, the
acquisition of the T. cruzi infections by blood
transfusion is reported; lately this route of the
infection acquisition is considered, carefully, as
well in several countries of the Northern
Hemisphere [34-36]. Occasionally, the T. cruzi
infections are acquired by accident in the
laboratory and by organ transplantation [36].
The transmission of the T. cruzi from the Chagas
mother to her offspring is an important route of
the infections in humans [36-38]. The congenital
transmission of Chagas disease refers to the in
uterus infection acquisition by the offspring.
However, pregnancy depends upon partnership
and male contribution to the in uterus T. cruzi
infections awaits investigation. In the course of
the pregnancy, the chagasic woman may
undergo early embryo resorption, miscarriage,
death birth, or neonatal death; live births can
either be T. cruzi-infected or infection-free,
healthy baby. Additionally, the infection-free,
healthy progeny can inherit the T. cruzi
mitochondrion minicircle kDNA sequences into
the genome [7].
3. CHAGAS DISEASE
The routes of acquisition of the T. cruzi infections
impose either the silent American
trypanosomiasis or the clinically manifested
Chagas disease. Fair accounts of the prevalence
of those patients are missing and, therefore, the
epidemiologic reports on morbidity and mortality
due to the T. cruzi infections are rather
inconclusive [6]. In the absence of disease
manifestation, chronically infected humans are
intermediate-phase carriers or reservoirs of the
T. cruzi infections [7,37,38]. A majority of
chronically infected individuals remains in the
intermediate phase and their life expectancy is
similar for non-infected individuals [7].
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
4
The field studies show that some chronically
infected individuals develop chronic Chagas
disease (CCD) usually over ~30 years after the
acquisition of the T. cruzi: 94.5% are cases
affected by heart trouble, among which 38.5%
die suddenly, and 56% succumb to heart failure
[6,7]. Sudden deaths are associated with
arrhythmias and heart rate turbulence. In
addition, megaesophagus and megacolon are
revealed in 5.5% of chronically infected people. A
gamut of neurologic syndromes and clinical
dysfunctions are manifestations of Chagas
disease, yet, scarcely studied [7-10]. Moreover,
CCD clinic and pathologic features, which are
indistinguishable from idiopathic inflammatory
dilated cardiomyopathy, have the differential
diagnosis set by the NAT-nDNA test [6,7].
4. DIAGNOSIS
4.1 Trypanosoma cruzi in the Blood
The direct microscopic demonstration of T. cruzi
in the blood is recorded in the early phase of the
infection. Also, the parasite is detected by
concentration methods, such as blood culture
and xenodiagnoses (having a clean bug to feed
upon the patient’s blood). For the epidemiologic
surveys, however, these diagnostics approaches
are out of reach by a majority of the T. cruzi
intermediate phase chronically infected
population, in a lack of late clinical CCD
manifestations.
4.2 Immune Diagnosis
In the absence of a direct demonstration of the T.
cruzi in patient’s blood the diagnosis relies on
searches for the specific antibody. The acutely
infected people are recognized by detecting IgM
antibody class or IgG serum conversion usually
after the second week of infection. Currently,
highly sensitive, but relatively less specific
indirect hemagglutination, enzyme-linked
immunosorbent assay (ELISA), and indirect
immunofluorescence (IFI) tests are routinely
used [41-42]. However, none of these assays
affords complete sensitivity and specificity,
because the host's immune system sees a
plethora of parasitic antigens and clonal selection
of immune responses [6,7,41,42]. A word of
caution is required because a grey zone of
inconclusive assays that yield cross-reactive
antibodies in the serum of patients with
Leishmania sp., Mycobacterium leprae and M.
tuberculosis, Treponema pallidum, or
autoimmune diseases [7,42]. Largely, the
employment of recombinant or synthetic peptide
antigens diminishes the sensitivity of the assays,
and these concerns bear practical consequences
[38,39]. With this respect, caution is required in
order to avoid an accidental T. cruzi-
contaminated blood transfusion. The health
systems should no longer rely solely on testing
the humoral immunity specific T. cruzi antibody
[7,41-47]. Nonetheless, the electrocardiograph
recordings in series of patients from endemic
regions show cases with the positive serum anti-
T. cruzi IgG, increased heart size in the chest X-
rays, and arrhythmias with features of chronic
Chagas disease [48-50].
4.3 Nucleic Acids Test (NAT)
The accuracy of the diagnoses of the T. cruzi
infections to the point-of-care for the assessment
and curtailment of the epidemics, and as well for
the health care delivery to thousands of people
aggravated by the yet incurable Chagas disease,
is fundamental [43-47,51,52]. Therefore, the
diagnosis of the sexually transmitted infection
requires the flagellate protozoa footprint [38-40,
43-47], because the specific IgG antibody is
absent in the immune tolerant progeny of the T.
cruzi-infected parents [38,51-54]. The immune
tolerance showed in the chicken model system
refractory to the T. cruzi infection after the 10
th
day of incubation [38,47,51-54]. The chicks
hatched from T. cruzi-inoculated eggs, which
grow to adult life, do not raise specific antibody
(Y immunoglobulin) after challenge with the
formalin killed parasite [53,54].
The NATs detect T. cruzi nuclear DNA (nDNA)
and its kinetoplast DNA (kDNA) in somatic cells,
and as well in germ cell line of the reproduction
systems immune privilege organs [38,43-47,51-
54]. Moreover, the NAT-nDNA test is an
indispensable condition for the diagnosis of T.
cruzi infection in immune-tolerant patients, in the
absence of the specific antibody. The
development of throughput digital PCR platforms
[47] needed to testing blood donor candidates,
so as deferral of those NAT-nDNA positive blood
donors, and to surveying accurate prevalence
and correct information on the epidemiology of
American trypanosomiasis and Chagas disease.
In so far, the NAT polymerase chain reaction
(PCR) is the basis for research laboratories
diagnosis of the T. cruzi infection and detection
of congenital Chagas disease at birth [38,43-46].
Its employment ensures tidy laboratory practices
to achieve a specific diagnosis of the highly
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
5
repetitive sequences in the flagellate protozoa,
showing homology with human chromosomes;
and throughput platform required for large-scale
epidemiologic surveys. A gamut of nDNA primer
sets is available for in-house PCR diagnosis of
the T. cruzi infections with variable sensitivity and
specificity [38, 44,47]. The Tcz1/2 primers set
that targets a 188-nt telomere DNA repeat in the
T. cruzi genome [48] are used in the family-
based studies [38-51]. The amplicons resolved in
agarose gels and transfer to positively charged
nylon membranes hybridize to the 188-nt α
32
p-
dATP labeled probe, subsequently subjected to
X-Rays film exposure during variable periods.
The positive amplicons are subjected to cloning
and sequencing. The bioinformatics analyses
reveal the 188-nt nDNA footprint in the blood and
in the semen samples. This in-house NAT is the
point-of-care diagnosis of an active T. cruzi
infection [38, 43-46]. Therefore, gold standard
NATs are required to guarantee integrity, purity,
and adequacy of blood bank supply donor
deferral and pathogen inactivation of blood
components. These efforts should be supported
by the health care networks for prevention of an
emerging T. cruzi threat; laboratory procedures
quality control assessment by skilled personnel
are needed to monitor all steps prior to the blood
transfusion and for delivery of the health care.
Additionally, the importance of the NAT tests for
diagnosis of the T. cruzi infection and CCD
cannot be underemphasized because the cross-
section epidemiological surveys based on the IFI
serological assessments of the specific antibody
determine neither the accurate prevalence of the
T. cruzi infections nor the ratios of the parasite
sexual transmission in the human population [38,
41-46].
5. TRANSFER OF KINETOPLAST DNA
(kDNA) FROM AMERICAN
TRYPANOSOMES TO HUMANS
The studies showed transfer of the mitochondrial
DNA (kDNA) minicircles sequences from
T. cruzi to the rabbit’s chromosome [56], and it
was further showed that the Chagas rabbit
progeny retained the protozoan minicircle
sequence mutation into the genome [7, 52-56].
The chicken family studies spurred the human
families’ investigations on the transfer of the
kDNA sequences from Chagas disease parents
to their progeny [54-56]. The founders of study
families showed kDNA minicircle sequences
integrated into the Long Interspersed Nuclear
Element-1 (LINE-1, equivalent to CR1 in the
chicken) of the human genome. In addition, the
kDNA minicircle sequences integrated at various
locations, such as structural, cell growth and
differentiation, immune responses and at
important biochemical pathways checkpoint
genes [52-56].
The development of Chagas heart trouble in the
chronically infected human population is an
ominous sign because the cases succumb to the
autoimmune inflammatory cardiomyopathy either
suddenly or in the range of six months to two
years after the beginning of clinical symptoms
and failure, and, therefore, it is unusual to find
those cases in a cross sectional field study.
Nevertheless, the family study showed the
accumulation of a minimum of 4 to 8 kDNA
mutations into the retrotransposable LINE-1
located at various chromosomes of CCD cases
with clinically manifested heart insufficiency [7,
38, 52-56].
The chagasic parental (F0) vertically transferred
the T. cruzi kDNA minicircle sequences to
the F1 and F2 progeny, which integrated
into the genome [38]. Additionally, the
families study population showed that
somatic cells had the T. cruzi NAT-nDNA positive
tests in family members and that sperm
donors had the active T. cruzi infection.
These findings suggest that intercourse is a
sustainable route for transmission of the T. cruzi
infection [38]. The clinic and epidemiologic
importance of these findings address to the
flagellate life-long infection, the overtime
accumulation of the mitochondrion kDNA
minicircle mutations and Chagas disease
multifaceted manifestations, in which parasite-
induced mutations underpinning the genetically
driven autoimmune pathogenesis, remarkably,
features of host-parasite relationships and
sprouting human disease.
6. SEXUAL TRANSMISSION OF THE
AMERICAN TRYPANOSOMES
In the last decades, the growing number of
Chagas disease in the Northern Hemisphere
inhabitants ever exposed to the insect vectors
suggests the possibility of acquisition of the T.
cruzi infections by the intercourse [7, 38]. In this
regard, the T. cruzi infections encrypted in
immune privilege body niches, in the absence of
inflammatory infiltrates, make the grounds for the
parasitic growth [57-63]
in the reproductive
organs (Fig. 1) and, thus expelling the protozoan
in the semen (Fig. 2) and in the uterine
secretions [38, 57].
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
6
6.1 Family Study
The timely demonstration of the sexually
transmitted T. cruzi in families’ document the
epidemic bursts of the infections in the
population that received health care at the
Center for Chagas Disease Surveillance. Among
individuals in the families, there were cases of
the acute T. cruzi infections showed by direct
demonstration of the blood trypomastigotes.
However, the epidemiologic history of the study
population revealed the absence of the
triatomine-bite transmission of the infections and,
also, the other routes of the infection-acquisitions
were denied.
In the family study, each acute case treated with
the anti-trypanosome nitro derivative
benznidazole (5 mg/kg/day) for 60 days, and all
family members received health assistance for
five years. The individuals in the families yielded
blood samples at 1, 2, and 3 years set points for
serum and DNA collection. Interestingly, the
NAT-nDNA test was consistently positive in 76%
independent assessments of blood samples
collected at three different set points: among
these, 28.4% tested IgG positive. This broad
discrepancy explained by the immune tolerance,
in the absence of the antibody in the majority of
NAT-kDNA positive individuals, since the
sexually transmitted T. cruzi could reach the
early embryo before the development of the
immune system [38,52-57]. Additionally, the
NAT-kDNA test was positive in 92.6% family
members (EMBL HG008116 to HG008708). The
difference between the results obtained by the
NAT-nDNA and the NAT-kDNA approaches
addresses to the mutations resulting from the
integration of the minicircle DNA sequences into
the human genome in the absence of live T. cruzi
infections. (Fig. 3).
In the absence of the T. cruzi antibody, the NAT
analyses of the germ cells collected from
subjects in the family study confirmed the
experimental data [38,52-56] showing that the T.
cruzi infections acquired during early embryo
growth do not bear the specific antibody,
whereas those acquired during fetal development
do not undergo immune-tolerance and, thus
reveal the specific antibody. These analyses
showed the T. cruzi nDNA- and kDNA-specific
bands in the somatic cells of the blood and in the
haploid semen samples examined [38, 57-63]
(Fig. 4). The data that suggest the sexual
transmission of the T. cruzi infections confirmed
by other experiment, in which the injection of 100
µl aliquot of NAT-nDNA positive Chagas patient’s
semen into the peritoneal cavity or its infusion
into the vagina of mice documented the infectivity
of the T. cruzi present in the human ejaculate:
one month thereafter, the growing T. cruzi
amastigote nests appeared in the heart and
skeletal muscles, and in the lumen of the vas
deferens and uterine tube (Fig. 4).
The early epidemiology based serological
surveys conducted in the spare individual
estimate that the prevalence of the T. cruzi
infections averages 7 ± 3 percent of the Latin
America population [6,49]. The family study NAT
assessments revealed much high ratios of the T.
cruzi infections in male and female family’s
founders and progeny mates [38,52-56].
However, the so-called congenital transmission
of the infection means that the woman only bears
for the vertical T. cruzi transmission to her baby.
Certainly, such prejudice definition is in the lack
of scientific understanding, because it omitted
the inevitable roles played by both mates during
the bi-partisan sexual reproduction [38]. With this
respect, further family-based studies at various
ecosystems are required to determine the role
played by the mate’s sexual intercourse in the
transmission of the infectious agent retained in
the immune privilege reproductive organs. The
immune privilege is the absence of destructive
inflammatory reactions to microbes’ antigen
loads in important body structures in the brain,
eyes, and reproductive organs, accomplishing
sensitive functions, reproduction, and the species
survival [64, 65]. The immune privilege
sets free
the T. cruzi growth in the organs of the
reproduction, whereas the immune tolerance [64]
prevents the immune system rejection of the
parasite antigen, because it is recognized as a
component of its own body since early embryo
life.
6.2 Mouse Model System
The pathology searches documented a gamut
of the T. cruzi amastigote nests in the
mouse uterine endometrium, ovary theca
cells, and testicles’ Sertoli cells and in the lumen
of the seminiferous tube, and in the epididymis,
seminal vesicles, prostate, and ureter [57-63].
Actually, the inoculation of the T. cruzi from the
hindgut of a triatomine bug incited the replicative
flagellates’ growth [57] in the recipient mouse
ejaculates (Fig. 2). Furthermore, the T. cruzi
infections were documented upon Chagas
patient’s semen inoculation or infusion in naïve
mice [38].
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
7
Fig. 1. The Trypanosoma cruzi infection in the seminiferous tube of a boy with the acute
Chagas disease. Notice the round amastigote forms in the gonia blasts and clumps of
amastigotes and trypomastigotes in the lumen of the tube (arrows). Bar, 10 µm.
Microphotograph from Doctor Antonio Teixeira’s file, 1970
Fig. 2. The Trypanosoma cruzi parasitic forms in the semen ejaculate of acutely infected mice.
The arrows indicate amastigote (A) within the head of a spermatozoan; free amastigotes (B)
and trypomastigotes (C and D); and the epimastigote forms (E and F) in the semen ejaculates
(arrows). Giemsa stained smears. Bars, 10 µm. Reprinted with permission from the Author and
the Publisher [57]
Fig. 3. The diagnosis of the Trypanosoma cruzi infections made by PCR and Southern
hybridizations. Notice the nuclear DNA (nDNA) bands in 11 Chagas patients with the live
infections, whereas 21 family members showed the mitochondrial DNA (kDNA) bands.
Reprinted with the permission from the Authors and the Publisher [38]
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
8
Fig. 4. The infectivity of the Trypanosoma cruzi from the human semen ejaculates upon aliquot
instills into the peritoneal or into the vagina of naïve mice. Left) the T. cruzi amastigotes clump
in the lumen of the vas deferens. Right) the T. cruzi amastigotes swarm epithelial cells of the
uterine tube (insert, dividing amastigotes). Bars, 10 µm. Reprinted with the permission from
the Authors and the Publisher [38]
Moreover, family studies about the T. cruzi-
infected male and female mice showed the
transmission of the protozoa flagellates to naïve
mates upon intercourse. In those experiments,
each breeding pair placed in one cage inside a
safe box avoided escaping. In experimental
group-A, 10 T. cruzi-infected males mated with
10 naive, control females. In experimental group-
B, 10 T. cruzi-infected females mated with 10
naive, control males [38]. After breeding, groups
A and B mates’ yielded NAT-nDNA bands that is,
the naive mates readily acquired the T. cruzi
after the sexual encounter. The triplicate groups
of independent experiments also showed that
naive female or male mouse that sexually mated
with a T. cruzi-infected male or female acquired
the infection. Those nDNA-positive founders (F0)
generated progeny F1 and F2 that they raised
until six weeks of age. The founders' (F0)
transmitted sexually the infections to F1 progeny
as shown by the nDNA-positive bands. The
breeding of these nDNA-positive mates
generated F2 progeny, which showed the nDNA
bands that indicate the vertically acquired
infections. In these experiments, 58.6% of the
progeny had the NAT-positive nDNA assays, and
22% of the progeny had anti-T. cruzi antibodies.
The results confirmed the absence of the specific
antibody in a majority of F1 and F2 mice (78%)
that bore vertical T. cruzi infections by
intercourse. After that, the tissue sections taken
for microscopic examination showed the T. cruzi
in the reproductive system of those mice [38]. In
summary, those experiments suggest that few
offspring (22%) of chagasic mates had the T.
cruzi-specific antibody, probably, because the
infectious agent reached the fetus with a mature
immune system.
7. DRUG DEVELOPMENT
The strategic for the treatment of Chagas
disease should take into consideration that single
infective T. cruzi initiates a lifelong infection [7].
Regardless of the route of the infection, after
several cycles of division, the T. cruzi ends up in
the reproduction organs immune privilege sites
where it grows [38,64]. The amazing capability of
the T. cruzi to thrive in the immune privilege
mammalian reproductive system, so as to reach
out the route to contaminate the host during
intercourse; the species survival prevail
transmission by sex and warrants the T. cruzi
anthropozoonosis, and, therefore, this new
concept shall guide the preventive medicine and
public health measures for the control of
American trypanosomiasis. This observation
makes the absolute need of the parasite
eradication in order to accomplish cure of
Chagas disease and prevention of the sexually
transmitted infections.
The lead drugs used for the treatment of the T.
cruzi infections are nitro- derivative compounds
nifurtimox [4-(5-nitro-phurylideneamino-)
tetrahydro-4-4-1, 4-thiazine-1-1-dioxide], and
benznidazole [N-benzyl-2-nitro-
imidazoleacetamide) [39-40, 49, 50, 81-84]. Both
drugs achieve elimination of the T. cruzi
infections in vitro but they fail to eradicate the live
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
9
infections in experimental animals and in man
[38,40,66,67], possibly, because after-treatment
non-phagocyte growing nest sustains the
persisting parasite infection. The nitro-derivatives
toxicity includes mutagenic, teratogenic,
carcinogenic and sterilizing activities [68-76],
which relies on the pteridine reductase activity
[85-90] upon the lead and release of high-energy
electron excited intermediates:
-
NO
2
,
-
HO,
1
O
2
,
H
2
O
2
that bind to macromolecules and DNA. The
drug toxicity targets the parasite and the
mammalian host cell [7-79].
8. NEW DRUGS FOR PREVENTING
CHAGAS DISEASE
The Chagas disease reported in five continents
[25-37] has brought much attention from private
and public organizations. In this new look,
searches for safe, efficacious, low-cost drugs to
curtailment of the epidemics needed.
Consistently, the T. cruzi biochemical pathways
checkpoints described herein suggest possible
targets for drug development. The clinical trials
with the 14α-demethylase (CYP51) that
catalyzes the removal of the 14 α-methyl group
from the scaffold revealed that posaconazole and
ravuconazole inhibitors of sterol biosynthesis
undergo treatment failure above that for
benznidazole-treated patients [38,43-50]. The
anti-trypanosome activity of lapachone-based
1,2,3-triazole naphthoquinones derivatives
presented IC50/24 h 80.8 values at 6.8 and 8.2
μM activity which is above that of the anti-T. cruzi
drug benznidazole. The nitro heterocycles
fexinidazole prompted the discovery of the
oxazole AN4169 (SCYX-6759) with anti-T. cruzi
activity in a mouse model. Other leads
derivatives of the herbicide fenarimol, which is an
azole inhibitor of T. cruzi CYP5128–30.
Meanwhile, the nitro derivatives benznidazole
and nifurtimox registered for treatment of Chagas
disease in Latin America [80-82]. In the countries
of the Northern hemisphere the prescription of
these drugs requires license from the health
authorities.
Actually, the sustainable search for compounds
from medicinal plants and drug development to
kill the T. cruzi is needed, because most
antibiotics derived from natural metabolites used
for defense, reproduction, and survival [82-83].
The use of throughput biotechnologies for the
systematic procurement and identification of
active molecules in plants from various
ecosystems, showing enormous biodiversity shall
enhance the chance for obtaining a top lead drug
without toxicity to eradicate the T. cruzi for
prevention and treatment of Chagas disease.
This is a milestone research endeavor.
9. PREVENTING AMERICAN
TRYPANOSOMIASIS AND CHAGAS
DISEASE
Over three decades the World Health Authorities
were at odds with the devastating acquired
immunodeficiency syndrome (AIDS). The
epidemics caused the death of unaccountable
people all over the world, in the lack of the
specific diagnosis, except for the variety of
opportunistic infections in the debilitating immune
tolerant organism. Otherwise, International
Health Authority Organizations consider Chagas
disease minor epidemics that affect the poor
people of Latin America, and the local Health
Institutions claim that the disease is under
control.
Meanwhile, the Chagas disease high morbidity
and mortality ratios are growing burden upon the
health systems worldwide and, therefore, it
should not be neglected anyway (21-37). On the
one hand, the Chagas disease unperceived
epidemics [84-85] require the attention of the
health systems because the potential of
spreading the re-emerging transmission routes,
including the sexually transmitted T. cruzi
infections can no longer be underemphasized.
On the other, the trophic network that enmeshes
the T. cruzi in countless ecotypes in different
ecosystems [4, 8], including the underground
[86], embracing many species of insect vectors,
most of which are not used to domiciliation are
not susceptible to insecticides, and, therefore,
the anthropozoonosis cannot be eliminated by
the vector control strategy. To approach the
magnificent problem the health personnel and
scientists agree that time-consuming,
unprecedentedly sustainable investigation
programs are necessary, in order to create the
knowledge new order to combat this threatening
epidemic, urgently.
9.1 Education Information and
Communication for Health (EICH)
The EICH program, aiming at the control of the
transmission of the T. cruzi infections, is needed.
The population is entitled to know about all
routes of transmission of the T. cruzi infections:
a) sexually transmitted; b) accidental
contamination of health care workers and of
research personnel, and through blood
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
10
transfusion not deferred by the antibody assays;
c) imprudent ingestion of contaminated food; d)
insect-vector transmitted T. cruzi infections,
which are limited to the American Continent. The
a) and b) modes of transmission of the infections
are prone to occur in the five continents. The
basic EICH program to prevent the transmission
of the silent T. cruzi infections shall take some
experience of from AIDS mass-media guidelines,
and the NAT nDNA exam performed early for the
detection of the two weeks period in which the
acute infections run in the absence of the specific
antibodies [38,87,88].
The EICH program should aim at the control of
the epidemics through the involvement of the
people in the community with the knowledge
about the trophic networks that associate the
insect vectors reservoirs with over one
thousands mammal hosts in the indomitable
nature [8]. The information about the several
dozens of the insect species (triatomine bugs)
transmitters of the T. cruzi, thriving in countless
ecotypes, hiding away under tree’s bark, in the
mammals and in the birds’ nests, as well under
the rocks, and in burrows in the undergrounds,
should be promptly delivered to the population [8,
11,17,18]. They should know that a few
triatomine bug species are susceptible to
domiciliation, and those are main targets for
elimination, to assure the family’s protection at
the household.
The EICH guidelines shall sustain the
knowledge, saying that the education tempers
the best health agent in the community.
Additionally, to inform the people in the
community about the genotoxicity of the nitro-
derivative pyrethroid pesticide sprayed in the
house regardless of the inhabiting infants and
adolescents [68,69]. In the case, the health
authorities believe it is necessary to spray the
pyrethroids an informed consent form with the
Family Leader signature is a prerequisite for the
insecticide spraying, in the household free of
infants and adolescents.
A formidable effort is required for developing the
EICH program aiming at the grass roots of the
populations under potential exposures to the T.
cruzi infections. Furthermore, the effective
prevention measures shall sustain the concept
that the EICH efficacy is a lot more protective of
the family in the household than the spray of
pesticides that do not reach most blood fasting
triatomines dispersed in the hideaway
environment. In addition, the citizen under the
benefits of the EICH program shall fight to
eliminate the nasty triatomine bug inside his
house.
The citizens shall be informed about the constant
increasing ratios of the T. cruzi infections in Latin
America and worldwide. The continuous exodus
of the people from Latin America to the Northern
Hemisphere partly explains the many cases of
the Chagas disease in the regions where the
insect-vector does not exist. Meanwhile, the
usefulness of the EICH program at school and at
social organizations, with emphasis on the
sociology, biological and health sciences, and
mass–media communication, shall prevail
worldwide in order to prevent the sexual
transmission of the infections [38,85]. Moreover,
unprecedented drug-discovery program needs
for the parasite eradication and prevention of the
sexually transmitted T. cruzi infections in the
human population.
10. DISCUSSION
The trypanosome survival in invertebrate and in
vertebrate animals include fine strategic
checkpoints such as hiding away underground
and association with countless ecotypes out of
reach of skilled health personnel and
insecticides. The plethora of countless factors,
playing important roles in the multi-factorial chain
of events related to transmission of the T. cruzi
infections requires further studies in the
laboratories and in the field. Meanwhile, the
complexity of the T. cruzi life-cycle does not
sustain the belief that currently used strategies
are good enough for preventing the T. cruzi
infections so as to safeguard the human
population [8]. Such epistemological definition is
essential because not yet defined cumbersome
factors have been brought into play by an
enormously complex chain of events and the T.
cruzi encroaches in major ecosystems. The
discovery of novel preventive and control
strategies should be further investigated by
scholars, before propagating the control of the T.
cruzi infection and Chagas disease is achieved.
Actually, the sexually transmitted T. cruzi
infection undergoes life-long encryption in the
body, and, in the absence of clinic
manifestations, the patients do not seek
preventive medical care. In the absence of a
drug to eradicate the T. cruzi infection, the EICH
program is the corner stone for preventing the
sexual transmission of the American
trypanosomiasis and Chagas disease.
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
11
11. CONCLUDING REMARKS
The EICH actions in the American Continent,
concerning the prevention of the triatomines from
close contact with the human population should
be conducted directly in communities,
elementary schools, churches and social clubs,
and reinforced by social marketing and mass
media communications. The population should
be promptly informed about the modes of
transmission of the T. cruzi infections that pose
daily threats to human health. A realistic and
affordable control of the sexually transmitted T.
cruzi infections and the curtailment of Chagas
disease rely on a robust EICH program.
The EICH preventive actions to halt the
American trypanosomyasis and Chagas disease
spread worldwide should be on the time table
because the sexual transmissions of the T. cruzi
infection run simultaneously with the transfer of
the parasitic kDNA to the human genome. This
frailty introduces a significant change in the
concept of public health, and preventive
medicine [7,38,67]. The silent parasite-induced,
genetically driven autoimmunity [38,64-66,67]
associates ubiquitous clinical manifestations with
high ratios of morbidity and mortality several
decades after the infection acquisition, and,
therefore, a conjunction of measures is
necessary to control the Chagas disease and its
potential to become pandemic:
1. Chagas disease is a global problem that
requires international solidarity, consortia,
and exchanges of expertise in order to
provide the means for protection of the
population under risks of acquisition of the T.
cruzi infections.
2. Education, Information, and Communication
mass-media program (EICH) undertaken,
soon is the best, in order to obtain full
compliance from the people’s community,
similar to that employed to control HIV-AIDS
epidemics.
3. Safe new drugs needed to eradicate the
cryptic T. cruzi infections: i) to treat all the T.
cruzi-infected people, so as to prevent the
sexual and the accidental modes of
transmission of the infections. ii) To arrest
the cardiomyopathy: pre-empt the severe
Chagas heart disease case, before the killing
of sick bone marrow progenitors of the
effector immune lymphocytes before healthy
bone marrow transplantation [64,65].
4. Development of throughput digital PCR
platforms for assessing the population under
risk of acquisition of the T. cruzi infections,
so as deferral of NAT-nDNA positive blood
donors.
5. The health personnel should share
responsibility with the community leaders
about the target-directed limited spraying of
the pyrethroid insecticide in the triatomine
bugs infested households.
ACKNOWLEDGEMENTS
This work was funded by the USA NIH grant R03
1164, and by the Brazilian Government
PRONEX/FAPDF/ MCT/ CNPq/CAPES grant
193.000.589/2009. The funders had no role in
study design, data collection and interpretation or
decision to submit the work for publication.
CONSENT
It is not applicable.
ETHICAL APPROVAL
It is not applicable.
COMPETING INTERESTS
The authors have declared no conflict of interest
exists.
REFERENCES
1. Lake JA, de la Cruz F, Ferreira PC, Morel
C, Simpson L. Evolution of parasitism:
kinetoplastid protozoan history
reconstructed from mitochondrial rRNA
gene sequences. Proc Natl. Acd. Sci. USA.
1988;85:4779-83.
2. Simpson AG, Lukes J, Roger AW. The
evolutionary history of kinetoplastids and
their kinetoplasts. Mol Biol Evol. 2002;
19:2071-6.
3. Lalonde RG, Holbein BE. Role of iron in
Trypanosoma cruzi infection of mice. J Clin
Invest. 1984;470:470-6.
4. Taylor MC, Kelly JM. Iron metabolism in
trypanosomatids, and its crucial role in
infection. Parasitol. 2010;137:899-917.
5. Coura JR. The main scenaries of Chagas
disease transmission. The vectors, blood
and oral transmissions. Mem. Inst.
Oswaldo Cruz. 2015;110:277-82.
6. Teixeira AR, Nascimento R, Sturm NR.
Evolution and pathology in Chagas
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
12
Disease. Mem Inst Oswaldo Cruz. 2006;
101:463-91.
7. Teixeira AR, Hecht MM, Guimaro MC,
Sousa AO, Nitz N. Pathogenesis of
Chagas disease: Parasite Persistence and
Autoimmunity. Clin Microbiol Rev. 2011;
24:502-630.
8. Teixeira AR, Monteiro SP, Rebelo JM,
Argañaraz ER, Vieira D, Lauria-Pires,
et
al. Emerging Chagas disease: Trophic
network and cycle of transmission of
Trypanosoma cruzi from palm trees in the
Amazon. Emerg Infect Dis. 2001;7:100
DOI: 10.3201/eid0701.700100
9. Chagas C. New human trypanosomiasis.
Morphology and lifecycle of
Schizotrypanum cruzi, the cause of a new
human disease. Mem Inst Oswaldo Cruz.
1909;1:59-218.
10. Chagas C. A new human disease.
Summary of etiological and clinical studies.
Mem Inst Oswaldo Cruz. 1911; 3:219-275.
11. Reisenman CE, Lawrence G, Guerenstein
PG, Gregory T, Dotson E, Hildebrand JG.
Infection of Kissing Bugs with
Trypanosoma cruzi, Tucson, Arizona,
USA. Emerg Infect Dis. 2010;16:400–5.
12. Klotz JH, Dorn PL, Logan JL, Stevens L,
Pinnas JL, Schmidt JO, et al. Kissing bugs:
Potential disease vectors and cause of
anaphylaxis. Clin Infect Dis. 2010;50:1629-
34.
13. Kjos SA, Dorn PL, Yabsley MJ, Kitron U,
Snowden KF, Logan KS, et al.
Identification of bloodmeal surces and
Trypanosoma cruzi infection in Triatomine
Bugs (Hemiptera: Reduviidae) from
residential settings in Texas, the United
States. J Med Entomol. 2013;50:1126-
1139.
14. Bussacos AC, Nakayasu ES, Hecht MM,
Assumpção TC, Parente JA, Soares CM,
et al. Redundancy of proteins in the
salivary glands of Panstrongylus megistus
secures prolonged procurement for blood
meals. J Proteomics. 2011;74:1693-700.
15. Moffitt JE, Venarske D, Goddard J, Yates
AB, de Shazo RD. Allergic reactions to
Triatoma bites. Ann Allergy Asthma
Immunol. 2003;91:122-8.
16. Hecht MM, Bussacos AC, Lozzi SP,
Santana JM, Teixeira AR. Triatoma
infestans chooses to feed upon immune
prey. Am J Trop Med Hyg. 2006;75:893-
900.
17. Martínez-Ibarra JA, Valencia-Navarro I,
León-Saucedo S, Ibáñez-Cervantes G,
Bustos-Saldaña R, et al. Distribution and
infection of triatomines (Hemiptera:
Reduviidae) by Trypanosoma cruzi in the
state of Michoacán, Mexico. Mem Inst
Oswaldo Cruz. 2011;106:445-50.
18. Schofield CJ. Trypanosoma cruzi - the
vector-parasite paradox. Mem Inst
Oswaldo Cruz. 2000;95:95:535-44.
19. Yoshida N. Molecular mechanisms of
Trypanosoma cruzi infection by oral route.
Mem Inst Oswaldo Cruz. 2009;104 Suppl
1:101-7.
20. Shikanai-Yasuda MA, Carvalho NB. Oral
transmission of chagas disease. Clin Infect
Dis. 2012;54:845-52.
21. Blumental S, Lambermont M, Heijmans C,
Rodenbach M-P, El Kenz H,
Sondag D, et
al. First documented transmission of
Trypanosoma cruzi infection through blood
transfusion in a child with sickle-cell
disease in Belgium. PLoS Negl Trop Dis.
2015;9:e0003986
DOI: 10.1371/journal.pntd.0003986
22. Ries J, Komarek A, Gottschalk J, Brand
B, Amsler L, Jutzi M, et al. A case of
possible chagas transmission by blood
transfusion in Switzerland. Transfuse.
Med. Hemother. 2016;43:415–417.
23. Dias JCP, Amato Neto V. Prevention
concerning the different alternative routes
for transmission of Trypanosoma cruzi in
Brazil. Rev Soc Bras Med Trop. 2011;44:
suppl 2:68-72.
24. Cevallos AM, Hernández R. Chagas’
disease: Pregnancy and Congenital
Transmission. 2014;ID 40186.
25. Jackson Y. Congenital transmission of
Chagas disease in Latin America. Emerg
Infect Dis. 2009;15:601-3.
26. Bern C, Kjos S, Yabsley M. Montgomery.
Trypanosoma cruzi and Chagas disease in
the United States. Clin Microbiol Rev.
2011;24:655-681.
27. Klein N, Hurwitz I, Durvasula R.
Globalization of chagas disease: A
Growing Concern in Nonendemic
Countries. Epidemo Res Int. 2012;ID
136793-806.
28. Liu Q, Zhou X-N. preventing the
transmission of american trypanosomiasis
and its spread into non-endemic countries.
Infect Dis Poverty. 2015;4:60-71.
29. Jackson Y, Pinto A, Pett S. Chagas
disease in Australia and New Zealand:
risks and needs for public health
interventions. Trop Med Int Health. 2014;
19:212-8.
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
13
30. Gascona J, Bernb C, Pinazoa M-J. Chagas
disease in Spain, the United States and
other non-endemic countries. Acta Tropica.
2010;115:22–27.
31. Bartoloni A, Zammarchi L. Chagas disease
in non-endemic countries, still a neglected
public health issue. Infectious Diseases
Unit, Department of Experimental &
Clinical Medicine; 2014. Florence, Italy.
Available:http://www.peah.it/2014/02/
32. Pérez-Molina JA, Norman F, López-Vélez
R. Chagas disease in non-endemic
countries: epidemiology, clinical
presentation and treatment. Curr Infect Dis
Rep. 2012;14:263-74.
33. Dorn PL, Perniciaro L, Yabsley MJ, Roellig
DM, Balsamo G, Diaz J, et al.
Autochthonous Transmission of
Trypanosoma cruzi, Louisiana. Emerging
Infect Dis. 2007;13:605-7.
34. Takeshi N, Miura S. Current situation of
chagas disease in non-endemic countries.
Junt Med J. 2015;61:389-95.
35. Albajar-Viñas P & Jannin J. The hidden
Chagas disease burden in Europe. Euro
Surveill. 2011;16:pii-19975.
36. Schmunis GA, Yadon ZE. Chagas disease:
a Latin American health problem becoming
a world health problem. Acta Trop. 2010;
115:14-21.
37. Castro AM, Vinaud M, Teixeira AR.
Emerging chagas disease. In: Chapter 1.
Chagas disease. A global Health Problem.
Bentham Science Publishers, New York;
2009.
38. Araujo PF, Almeida AB, Pimentel FC, Rios
A, Sousa A, Valente SA, Valente VC, et al.
Sexual transmission of american
trypanosomes in humans. New potential to
become pandemic. Mem Inst Oswaldo
Cruz. 2017;112:1-10.
39. Tarleton RL, Gürtler RE, Urbina JA,
Ramsey J, Viotti R. Chagas Disease and
the London Declaration on Neglected
Tropical Diseases. PLoS Negl Trop Dis.
2014;8:e3219.
DOI:10.1371/journal.pntd.0003219 PMCID
PMC4191937.
40. Braga MS, Lauria-Pires L, Nitz N, Rosa
AC, Argañaraz ER. The treatment of
Chagas disease patients with nitro
derivative is unsatisfactory. Rev Inst Med
Trop São Paulo. 2001;43:175-181.
DOI:org/10.1590/S0036-
46652001000300013
41. Teixeira AR, Vexenat A. The truthful
meaning of immunologic exams used in
the diagnosis of endemic diseases. Rev
Soc Bras Med Trop. 1996;29:379-82.
42. Vexenat AC, Santana JM, Teixeira, ARL.
Cross-reactivity of antibodies in human
infections by the kinetoplastid protozoa
Trypanosoma cruzi, Leishmania chagasi
and Leishmania (Viannia) braziliensis. Rev
Inst Med Trop São Paulo. 1996;38:177-85.
43. Britto CC. Usefulness of pcr-based assays
to assess drug efficacy in chagas disease
chemotherapy: value and limitations. Mem
Inst Oswaldo Cruz. 2009;104(suppl 1):122-
35.
44. Hsieh H, Patterson AS, Ferguson BS,
Plaxco KW, Soh HT. Rapid, Sensitive, and
quantitative detection of pathogens DNA at
the point-of-care via microfluidic
electrochemical quantitative loop-mediated
isothermal amplification (MEQ-LAMP).
Angew Chem Int Ed Engl. 2012;51:4896-
900
DOI: 101002/anie.201109115
45. Virreira M, Truyens C, Alonso-Vega C,
Brutus L, Jijena J, Torrico F. Comparison
of Trypanosoma cruzi lineages and levels
of parasitic DNA in infected mothers and
their newborns. Am J Trop Med Hyg. 2007;
77:102-106.
PMID: 17620638
46. Mendes DG, Lauria-Pires L, Nitz N, Lozzi
SP, Nascimento RJ, Monteiro PS, et al.
Exposure to mixed asymptomatic
infections with Trypanosoma cruzi,
Leishmania brazilieinsis and Leishmania
chagasi in the human population of the
greater Amazon. Trop Med Int Health.
2007;12:629-36.
47. Troger V, Nieman K, Gartig C, Kulhlmeir D.
Isothermal amplification and quantitation of
nucleic acids and its use in mycrosystems.
Nonomed. Nanotechnol. 2015;6:282-3.
48. Teixeira AR, Nitz N, Guimaro MC, Gomes
C, Santos-Buch CA. Chagas disease.
Postgrad Med J. 2006;82:788-95.
49. World Health Organization. Control:
second report of a WHO Expert Committe.
World Health Organ Tech Rep Ser. 2002;
905:1-109.
50. Balouz V, Aguero F, Buscaglia CA.
Chagas disease diagnostic applications:
present knowledge and future steps. Adv
Parasitol. 2017;97:1-45.
51. Moser DR, Kirchhoff LV, Sonwlaon JE.
Detection of Trypanosoma cruzi by DNA
amplification using the polymerase chain
reaction. J Clin Microbiol. 1989;27:1477-
82.
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
14
52. Teixeira AR, Gomes C, Nitz N, Sousa AO,
Alves RM, Guimaro MC, et al.
Trypanosoma cruzi in the Chicken Model:
Chagas-Like Heart Disease in the Absence
of Parasitism. PLoS Negl Trop Dis. 2011;
5:e1000.
53. Guimaro MC, Alves RM, Rose E, Sousa
AO, Rosa AC, Hecht MM, et al. Inhibition
of autoimmune chagas-like heart disease
by bone marrow transplantation. PLoS
Negl Trop Dis. 2014;8:e3384.
54. Nitz N, Gomes C, Rosa AC, D’Souza-Ault
MR, Moreno F, Lauria-Pires L, et al.
Heritable integration of kDNA Minicircle
Sequences from Trypanosoma cruzi into
the Avian Genome: Insights into Human
Chagas Disease. Cell. 2004;118:175–186.
55. Hecht MM, Nitz N, Araujo PF, Sousa AO.
Rosa AC, Gomes DA, et al. Inheritance
of DNA transferred from American
trypanosomes to human hosts. PLoS One.
2010;12:e9181.
56. Hecht MM, Nitz N, Araujo PF, Teixeira AR.
Pathogenesis of Chagas disease. Lateral
DNA transfer, genotype alterations and
autoimmunity. Rev Soc Bras Med Trop.
2009;4(Supl 1):68-72.
57. Alarcón M, Moreno E, Colasante C,
Yarbuh AL, Cáceres K, Araujo S.
Presencia de epimastigotes de
Trypanosoma cruzi en el plasma seminal
de ratones con infección aguda. Bol Mal
Salud Amb. 2011;51:237.
58. Lenzi HL, Castelo-Branco MT, Pelajo-
Machado M, Oliveira DN, Gattass CR.
Trypanosoma cruzi: compromise of
reproductive system in acute murine
infection. Acta Trop. 1998;71:117.
59. Carvalho LO, Abreu-Silva AL, Hardoim D
de J, Tedesco RC, Mendes VG, Gonçalves
da Costa SC et al. Trypanosoma cruzi and
myoid cells from seminiferous tubules:
Interaction and relation with fibrous
components of extra cellular matrix in
experimental Chagas disease. Int J Exp
Path. 2009;90:52.
60. A Rios, M Ribeiro, A Sousa, F Pimentel, L
Hagström, R Andrade, et al. Can sexual
transmission support the enzootic cycle of
Trypanosoma cruzi? Mem Inst Oswaldo
Cruz, Rio de Janeiro. 2018;113(1):3-8.
61. Ribeiro M, Nitz N, Santana C, Moraes A,
Hagström L, Andrade R, et al. Sexual
transmission of Trypanosoma cruzi in
murine model. Exp Parasitol. 2016;162:
1-6.
DOI: 10.1016/j.exppara.2015.12.017.
62. Martin DL, Lowe KR, McNeill T, Thiele EA,
Roellig DM, Zajdowicz J, et al. Potential
sexual transmission of Trypanosoma cruzi
in mice. Acta Trop. 2015;149:15-8.
DOI: 10.1016/j.actatropica.2015.05.002.
63. Freitas JM, Pinto LA, Pimenta JR,
Rodrigues LB, Gonçalves VF Teixeira SM.
Ancestral Genomes, Sex, and the
Population Structure of Trypanosoma cruzi
PLoS Path; 2006.
Available:https://doi.org/10.1371/journal.pp
at.0020024
64. Fujisaki J, Wu J, Carlson AL, Silberstein L,
Putheti P, Larocca R, et al. In vivo imaging
of Treg cells providing immune privilege to
the haematopoietic stem-cell niche. Nature.
2011;474:216-219.
65. Schwartz RH. Historical overview of
immunological tolerance. Cold Spring Harb
Perspect Biol. 2012;4:a006908.
66. Lauria-Pires L, Braga MS, Vexenat AC,
Nitz N, Simoões-Barbosa A, Tinoco DL, et
al. Progressive chronic Chagas heart
disease ten years after treatment with anti-
Trypanosoma nitro derivatives. Am J Trop
Med Hyg. 2000;63:111-118.
PMID: 11388500.
67. Morillo CA, Marin-Neto JA, Avezum A,
Sosa-Estani S, Rassi-Jr A. Randomized
Trial of Benznidazole for Chronic Chagas'
Cardiomyopathy. N Engl J Med. 2015;
373:1295-1306.
DOI: 10.1056/NEJMoa15075742015.
68. Teixeira AR, Silva R, Rizzo LV, Cunha
neto E. Trypanocidal nitroarene treatment
of experimental Trypanosoma cruzi
infection does not prevent progression of
chronic-phase heart lesions in rabbits. J
Infect Dis. 1990;162:1420.
69. Teixeira AR, Cordoba JC, Souto Maior I,
Solorzano E. Chagas disease: Lymphoma
growth in rabbits treated with
benznidazole. Am J Trop Med Hyg. 1990;
43:146-158.
70. Teixeira AR, Jabur E, Cordoba JC, Mayor
ICS, Solorzano E. Alteração da resposta
imune mediada por células durante o
tratamento com Benzonidazol. Rev Soc
Bra Med Trop. 1983;16:11-22.
71. Teixeira AR, Silva R, Cunha Neto E, Rizzo
LV, Santana JM. Malignant, non-hodgkin's
lymphomas in Trypanosoma cruzi-infected
rabbits treated with nitroarenes. J Comp
Pathol. 1990;103:37-48.
72. Teixeira A, Calixto M, Teixeira M. Chagas'
disease: Carcinogenic activity of the
Teixeira et al.; ISRR, 7(2): 1-15, 2018; Article no.ISRR.42594
15
antitrypanosomal nitroarenes in mice.
Mutat Res. 1994;305:189-96.
73. Teixeira AR, Resende Filho J, Figueiredo
FC. A clinical, immunological and
pathological study in rabbits. Am J Trop
Med Hyg. 1983;32:258-272.
74. Moreno SN, Docampo R. Mechanism of
toxicity of nitro compounds used in the
chemotherapy of trichomoniasis. Environ
Health Perspect. 1985;64:199–208.
PMID: 3830698.
75. Miller R, Buttigan BE. Role of oxidants in
microbial pathophysiology. Clin Microbiol
Rev. 1997;10(1):1-18.
76. Traub-Cseko YM, Ramalho-Ortigão JM,
Dantas AP, de Castro SL, Barbosa HS,
Downing KH. Dinitroaniline herbicides
against protozoan parasites. The case of
Trypanosoma cruzi. Trends Parasitol.
2001;17(3):136-141.
77. Dickinson BC, Chang CJ. Chemistry and
biology of reactive oxygen species in
signaling or stress responses. Nature
Chem Biol. 2011;7:504–511.
DOI: 10.1038/nchembio.607.
78. Vermes A, Guchelaar HJ, Dankert DJ.
Flucitosine: A reviews of its pharmacology,
clinical indications, pharmaco-kinetics,
toxicity and drug interactions. J Antimicrob
Chemoth. 2000;46:171-179.
DOI: 10.1093/jac/46.2.171.
79. Augusto LS, Moretti NS, TCP
Ramos, Jesus TCL, Zhang M, Castilho BA,
et al. A membrane-bound eIF2 Alpha
Kinase Located in Endosomes Is
Regulated by Heme and Controls
Differentiation and ROS Levels in
Trypanosoma cruzi. PLoS Pathol. 2015;
11:e1004618.
DOI: 10.1371/journal.ppat.1004618.
80. Buckner SB, Navabi N. Advances in
Chagas disease drug development. Curr
Opin Infect Dis. 2010;23:609-616.
81. Chatelain E. Chagas disease drug
discovery. Toward a New Era. J Biomol
Screen. 2015;20:22-35.
82. Weniger B. Robledo S, Arango GJ, Deharo
E, Aragón R. Antiprotozoal activities of
Colombian plants. J Ethnopharmacol.
2001;78:193-200.
83. Bern C, Kjos S, Yabsley MJ. Trypanosoma
cruzi and chagas disease in the United
States. Clin Microbiol Rev. 2011;24:655-
681.
84. 84. Abad-Franck F, Gurgel-Gonçalves R,
Gurtle RE. On bugs and bias: Improving
Chagas disease control assessment. Mem
Inst Oswaldo Cruz. 2014;109:125-130.
85. Hotez PJ, Dumonteil E, Woc-Colburn L,
Serpa JA, Bezek S. Chagas disease: The
new HIV/AIDS of the Americas. Plos Negl
Trop Dis. 2012;6:e1498.
86. Lövy M, Šklíba J, Hrouzková E, Dvořáková
V, Nevo E. Habitat and burrow system
characteristics of the blind mole rat Spalax
galili in an area of supposed sympatric
speciation. PLoS One. 2015;10:e0133157.
87. Lloyd LS. Environmental Health Project
Activity Report nº 56. An assessment of
health education materials for control of
dengue and malaria in Honduras. USAID,
Arlington, Virginia Environmental Health;
1999.
88. Williams-Blangero S, VandeBerg J,
Teixeira AR. Attitudes towards Chagas
disease: Working to overcome the global
impact of neglected tropical diseases. Cad
Saude Publ. 1999;15:7-13.
_______________________________________________________________________________
© 2018 Teixeira et al.; This is an Open Access article distributed under the terms of the Creative Commons Attribution
License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any
medium, provided the original work is properly cited.
Peer-review history:
The peer review history for this paper can be accessed here:
http://www.sciencedomain.org/review-history/25651
... En Colombia, la prevalencia de esta enfermedad se ha estimado entre 700 000 y 1 200 000 habitantes de infectados, que habitan en 539 municipios de 15 departamentos del país en áreas de alto, medio y bajo riesgo, incrementada por los brotes de trasmisión oral de la infección.(3) A pesar de los programas de control de vectores, manejo de la tamización en población en riesgo además de gran impacto en el conocimiento del parásito T. cruzi, mediante investigación en relación al vector, los mecanismos de transmisión y la inmunología, esta enfermedad sigue siendo un problema de salud emergente en áreas no endémicas debido a los movimientos poblacionales y en áreas endémicas, en parte, por la falta de conocimiento sobre la enfermedad, la carencia de información y las acciones que toma la población frente a factores epidemiológicos o presentación sintomática probable de la enfermedad, lo cual además varía en cada ámbito social, debido a la diversidad cultural.(4) La articulación de un instrumento que se acerque al análisis del paradigma de la enfermedad de Chagas, mediante la perspectiva teórica como se proyecta en los procesos de validación,(5) permitirá un acercamiento a la realidad desde el punto de vista de la población frente a la problemática de la enfermedad de Chagas, que de alguna manera oriente el establecimiento de políticas que impacten la reducción de la morbimortalidad que ocasiona.Además, permitirá obtener un conocimiento válido generado a través de la investigación sistemática.(6) ...
Article
Full-text available
Introducción: La enfermedad de Chagas es de alta endemia en Colombia y es considerada un problema de salud pública por la morbilidad y mortalidad que ocasiona. Conocer la percepción de las comunidades frente a esta enfermedad es relevante en cuanto a las estrategias oportunas que se pueden establecer para su prevención. Objetivo: Evaluar la fiabilidad y validez de un instrumento que permita medir los conocimientos, actitudes y prácticas frente a la enfermedad de Chagas en comunidades endémicas. Métodos: Se desarrolló un estudio de corte transversal para la validación del contenido de un instrumento, diseñado con base en la literatura, el cual se sometió a la valoración por un panel de expertos que determinaron la eficiencia, relevancia, claridad y coherencia de cada uno de los ítems. Se determinó el índice Kappa de Randolph para evaluar concordancia. Luego se aplicó a 135 personas de áreas endémicas y se estableció el alfa de Cronbach para determinar consistencia interna. Resultados: El instrumento estudiado demostró concordancia según el índice de Randolph con una Kappa entre 0,7-0,8 para los ítems evaluados, con un alfa de Cronbach de 0,855 lo cual denota una consistencia adecuada. El cuestionario finalmente se estructuró con un total de 27 ítems que abordan: factores epidemiológicos y conocimientos de la enfermedad junto con actitudes y prácticas frente a esta. Conclusiones: El instrumento tiene un nivel adecuado de consistencia interna y concordancia, que permitiría su aplicación en áreas endémicas con poblaciones en riesgo a adquirir la enfermedad de Chagas.
... American trypanosomiasis, also known as Chagas disease, is endemic to people in rural areas of South America, where the Trypanosoma cruzi parasites are sympatric to the hematophagous triatomine insect vectors (Prata, 2001;Coura and Viñas, 2010;Teixeira et al., 2018). The migration of T. cruzi-infected people to the Northern Hemisphere and of travelers to endemic countries have likely made American trypanosomiasis a global challenge, since these infections can be transmitted from mother to offspring (Murcia et al., 2013), by blood transfusion and donated organs, and by contamination of hospital and laboratory workers (Teixeira et al., 2011a). ...
Article
Full-text available
Objective To contribute to the discussion on the research findings indicating the sexual transmission of American trypanosomiasis and Chagas disease in humans. Methods A review of the literature was performed to investigate the routes of transmission of Trypanosoma cruzi parasites and to evaluate the distribution of Chagas disease, which is now found across five continents. Results The epidemiological profile of American trypanosomiasis, which is still considered a neglected disease of the poor people of Latin America, has changed over time. A family-based study demonstrated that the blood protozoan T. cruzi can be transmitted sexually from infected males and females to naïve mates. Conclusions Evidence that Chagas disease can be transmitted sexually, coupled with the migration of individuals with Chagas disease to previously non-endemic countries and increased travel to endemic countries, has implications for public health. Improved screening of blood supplies and prenatal care are required to prevent congenital spread.
Article
Glucokinase (GLK) and Hexokinase (HK) have been characterized as essential targets in Trypanosoma cruzi ( Tc )‐mediated infection. A recent study reported the propensity of the concomitant inhibition of Tc GLK and Tc HK by compounds GLK2‐003 and GLK2‐004, thereby presenting an efficient approach in Chagas disease treatment. We investigated this possibility using atomic and molecular scaling methods. Sequence alignment of Tc GLK and Tc HK revealed that both proteins shared approximately 33.3% homology in their glucose/inhibitor binding sites. The total binding free energies of GLK2‐003 and GLK2‐004 were favorable in both proteins. PRO92 and THR185 were pivotal to the binding and stabilization of the ligands in Tc GLK, likewise their conserved counterparts, PRO163 and THR237 in Tc HK. Both compounds also induced a similar pattern of perturbations in both Tc GLK and Tc HK secondary structure. Findings from this study therefore provide insights into the underlying mechanisms of dual inhibition exhibited by the compounds. These results can pave way to discover and optimize novel dual Tc inhibitors with favorable pharmacokinetics properties eventuating in the mitigation of Chagas disease.
Article
We investigated the immunomodulatory, antiparasitic and cardioprotective effects of a sesquiterpene lactone (SL) administered alone or combined with benznidazole (Bz), in a murine model of Chagas' disease by in vitro and in vivo assays. Antiparasitic and cytotoxic potential of tagitinin C (SL) and Bz were tested in vitro against T. cruzi epimastigotes and cardiomyocytes. Swiss mice challenged with T. cruzi were also treated for 20 days with tagitinin C (10 mg/kg) alone and combined with Bz (100 mg/kg). Tagitinin C exhibited a higher antiparasitic (IC50: 1.15 µM) and cytotoxic (CC50 at 6.54 µM) potential than Bz (IC50: 35.81 µM and CC50: 713.5 µM, respectively). When combined, these drugs presented an addictive interaction, determining complete suppression of parasitemia and parasitological cure in all infected mice (100%) compared to those receiving Bz alone (70%). Anti-T. cruzi immunoglobulin G, and pro-inflammatory cytokines IFN-γ and TNF-α levels were reduced in animals treated with tagitinin C combined with Bz, while IL-10 production was unaffected. Heart inflammation was undetectable in 90% of the animals receiving this combination, while only 50% of the animals receiving Bz alone showed no evidence of myocarditis. Together, our findings indicated that the combination of tagitinin C and Bz exerts potent antiparasitic, immunomodulatory and cardioprotective effects. Due to the remarkable suppression of parasitemia and high parasitological cure, this combination was superior to Bz monotherapy, indicating a high potential for the treatment of Chagas's disease.
Article
Full-text available
Introduction: Disclaim of the Chagas disease case in Blood Center is based on the immunofluorescence and enzyme-linked immunosorbent antibody assays. Methods: The antibody assays were conducted in parallel with the nuclear DNA-PCR test in blood samples of 145 families’ members. Result: The specific antibody tests were positive in 26.9% (39/145) subjects. Contrastingly, the Trypanosoma cruzi nuclear DNA-PCR was positive in 72.4% (105/145) of those blood samples. Conclusion: The immune tolerance herein explains the reported discrepancies of results between the specific antibody and the accurate DNA analysis. The warranty of safe blood transfusion is achieved by the PCR technology.
Article
Full-text available
Background: Trypanosoma cruzi circulates in sylvatic habitats, mainly through blood-feeding triatomines, although other routes also contribute to its dispersion. Sexual transmission of T. cruzi is an understudied topic, especially among wild mammals. Because of the difficulties inherent to field work, experimentally infected mice are frequently used to evaluate the transmission of T. cruzi. Objective: This study aimed to evaluate the sexual transmission of T. cruzi in acutely infected mice. Methods: Male and female mice in the acute phase of Chagas disease were mated with naïve partners. Then, parasitological tests, immunohistochemistry, serological assays, and polymerase chain reaction (PCR) assays were used to detect infection. Findings: Parasitological analysis showed trypomastigotes in the blood of 20% of the naïve mice after mating with infected partners. Serological assays detected anti-T. cruzi antibodies in all naïve females mated with infected males and in 60% of naïve males mated with infected females. PCR showed T. cruzi nDNA bands for all naïve mice mated with infected partners. The possibility of sexual transmission was also confirmed by visualisation of amastigotes in the testes. Main conclusions: Our results demonstrate that sexual transmission of T. cruzi is an ordinary event that may contribute to maintenance of the parasite's enzootic cycle.
Article
Full-text available
Background: Transfusion-transmitted Chagas disease has been reported from endemic countries in Latin America. Switzerland is a non-endemic country but high prevalence of antibodies against Trypanosoma cruzi was found among immigrants. Immigrants may participate in blood donation; therefore, risk-adapted anti-T. cruzi screening for blood donors was implemented in Switzerland in 2013. Methods: Between January 2013 and July 2015, 1 out of 1,183 at-risk donors, tested at Blood Transfusion Service Zurich, was found anti-T. cruzi IgG-positive. Results and conclusion: Out of 54 donations given by the index donor (ID), we identified 77 blood products which were delivered to hospitals. Archived serum samples from the donations given during the prior 5 years were available for retrospective testing. All samples from ID revealed positive findings for anti-T. cruzi IgG. Donor-triggered look-back procedure identified a 70-year-old male recipient of a platelet concentrate (PC) donated by ID. The recipient succumbed of acute T. cruzi infection 2 years after transfusion of the PC.
Article
Full-text available
American trypanosomiasis, commonly known as Chagas disease, is caused by the flagellate protozoan parasite Trypanosoma cruzi. An estimated eight million people infected with T. cruzi currently reside in the endemic regions of Latin America. However, as the disease has now been imported into many non-endemic countries outside of Latin America, it has become a global health issue. We reviewed the transmission patterns and current status of disease spread pertaining to American trypanosomiasis at the global level, as well as recent advances in research. Based on an analysis of the gaps in American trypanosomiasis control, we put forward future research priorities that must be implemented to stop the global spread of the disease. Electronic supplementary material The online version of this article (doi:10.1186/s40249-015-0092-7) contains supplementary material, which is available to authorized users.
Article
Full-text available
Chagas disease is now a public health problem due to the detection of cases all around the world. The migration of the Latin American populations to North America, Europe, Asia and Oceania has made the parasite cross the seas, so as to secure the Trypanosoma cruzi survival by means of blood transfusion and by congenital transmission from mother to offspring. The increasing number of cases of Chagas disease detected in various countries all over the world urges the health systems to be prepared to its proper diagnosis and treatment.
Article
Full-text available
A costly search for food in subterranean rodents resulted in various adaptations improving their foraging success under given ecological conditions. In Spalax ehrenbergi superspe-cies, adaptations to local ecological conditions can promote speciation, which was recently supposed to occur even in sympatry at sites where two soil types of contrasting characteristics abut each other. Quantitative description of ecological conditions in such a site has been, nevertheless, missing. We measured characteristics of food supply and soil within 16 home ranges of blind mole rats Spalax galili in an area subdivided into two parts formed by basaltic soil and pale rendzina. We also mapped nine complete mole rat burrow systems to compare burrowing patterns between the soil types. Basaltic soil had a higher food supply and was harder than rendzina even under higher moisture content and lower bulk density. Population density of mole rats was five-times lower in rendzina, possibly due to the lower food supply and higher cover of Sarcopoterium shrubs which seem to be avoided by mole rats. A combination of food supply and soil parameters probably influences burrowing patterns resulting in shorter and more complex burrow systems in basaltic soil.
Article
Full-text available
Infection with the protozoan parasite Trypanosoma cruzi, the etiologic agent of human Chagas disease, results in life-long infection. Infective trypomastigotes circulate in the bloodstream and have the capacity to infect any cell type, including reproductive tissue. This study sought to assess the potential for sexual transmission of T. cruzi in an experimental mouse model. The conditions used in this study, in which acutely infected males and immunosuppressing the females, created a worst-case scenario allowing for the greatest chance of measuring transmission through intercourse. Male BALB/c mice were infected and mated with uninfected females, and the females were subsequently examined for T. cruzi tissue parasitism. A single transmission event of 61 total matings was observed, indicating a low but non-zero risk potential for male-to-female sexual transmission of T. cruzi. Copyright © 2015. Published by Elsevier B.V.
Article
Full-text available
Translation initiation has been described as a key step for the control of growth and differentiation of several protozoan parasites in response to environmental changes. This occurs by the activation of protein kinases that phosphorylate the alpha subunit of the translation initiation factor 2 (eIF2α), which decreases translation, and in higher eukaryotes favors the expression of stress remedial response genes. However, very little is known about the signals that activate eIF2α kinases in protozoan parasites. Here, we characterized an eIF2α kinase of Trypanosoma cruzi (TcK2), the agent of Chagas' disease, as a transmembrane protein located in organelles that accumulate nutrients in proliferating parasite forms. We found that heme binds specifically to the catalytic domain of the kinase, inhibiting its activity. In the absence of heme, TcK2 is activated, arresting cell growth and inducing differentiation of proliferative into infective and non-proliferative forms. Parasites lacking TcK2 lose this differentiation capacity and heme is not stored in reserve organelles, remaining in the cytosol. TcK2 null cells display growth deficiencies, accumulating hydrogen peroxide that drives the generation of reactive oxygen species. The augmented level of hydrogen peroxide occurs as a consequence of increased superoxide dismutase activity and decreased peroxide activity. These phenotypes could be reverted by the re-expression of the wild type but not of a TcK2 dead mutant. These findings indicate that heme is a key factor for the growth control and differentiation through regulation of an unusual type of eIF2α kinase in T. cruzi.
Article
Chagas disease, or American trypanosomiasis, is caused by the parasitic protist Trypanosoma cruzi. 10 million people are estimated to live with this disease. Chagas disease is endemic to the Americas, which corresponds to the distribution of the insect vectors, blood-sucking triatomine bugs. The presence of many mammalian species as reservoir hosts and the occurrence of a long asymptomatic phase of infection that may last more than 10 years make control difficult. In the United States, domestic transmissions from triatomines to humans are rarely reported, but it is estimated that there are 300,000 people living with Chagas disease among Latin American immigrants. Patients with Chagas disease are also found outside the Americas, as well as in Japan, via international migration. Thus, there is a growing need to understand the current situation in non-endemic countries in terms of establishing better preparedness against the incursion of Chagas disease.